Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38542109

ABSTRACT

The combination of signals from the T-cell receptor (TCR) and co-stimulatory molecules triggers transcriptional programs that lead to proliferation, cytokine secretion, and effector functions. We compared the impact of engaging the TCR with CD28 and/or CD43 at different time points relative to TCR engagement on T-cell function. TCR and CD43 simultaneous engagement resulted in higher CD69 and PD-1 expression levels than in TCR and CD28-stimulated cells, with a cytokine signature of mostly effector, inflammatory, and regulatory cytokines, while TCR and CD28-activated cells secreted all categories of cytokines, including stimulatory cytokines. Furthermore, the timing of CD43 engagement relative to TCR ligation, and to a lesser degree that of CD28, resulted in distinct patterns of expression of cytokines, chemokines, and growth factors. Complete cell activation was observed when CD28 or CD43 were engaged simultaneously with or before the TCR, but ligating the TCR before CD43 or CD28 failed to complete a cell activation program regarding cytokine secretion. As the order in which CD43 or CD28 and the TCR were engaged resulted in different combinations of cytokines that shape distinct T-cell immune programs, we analyzed their upstream sequences to assess whether the combinations of cytokines were associated with different sets of regulatory elements. We found that the order in which the TCR and CD28 or CD43 are engaged predicts the recruitment of specific sets of chromatin remodelers and TFSS, which ultimately regulate T-cell polarization and plasticity. Our data underscore that the combination of co-stimulatory molecules and the time when they are engaged relative to the TCR can change the cell differentiation program.


Subject(s)
CD28 Antigens , Receptors, Antigen, T-Cell , CD28 Antigens/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes , Lymphocyte Activation , Cell Differentiation , Cytokines/metabolism
2.
Int J Inflam ; 2024: 2205864, 2024.
Article in English | MEDLINE | ID: mdl-38250663

ABSTRACT

Inflammatory and antimicrobial diseases constitute a major burden for society, and fighting them is a WHO strategic priority. Most of the treatments available to fight inflammatory diseases are anti-inflammatory drugs, such as corticosteroids or immunomodulators that lack cellular specificity and lead to numerous side effects. In addition to suppressing undesired inflammation and reducing disease progression, these drugs lessen the immune system protective functions. Furthermore, treating infectious diseases is more and more challenging due to the rise of microbial resistance to antimicrobial drugs. Thus, controlling the inflammatory process locally without compromising the ability to combat infections is an essential feature in the treatment of inflammatory diseases. We isolated three forms (DRS-DA2N, DRS-DA2NE, and DRS-DA2NEQ) of the same peptide, DRS-DA2, which belongs to the dermaseptin family, from the Mexican tree frog Pachymedusa dacnicolor. Interestingly, DRS-DA2N and DRS-DA2NEQ exhibit a dual activity by inducing the death of leukocytes as well as that of Gram-negative and Gram-positive bacteria, including multiresistant strains, without affecting other cells such as epithelial cells or erythrocytes. We showed that the death of both immune cells and bacteria is induced rapidly by DRS-DA2 and that the membrane is permeabilized, leading to the loss of membrane integrity. We also validated the capacity of DRS-DA2 to regulate the pool of inflammatory cells in vivo in a mouse model of noninfectious peritonitis. After the induction of peritonitis, a local injection of DRS-DA2N could decrease the number of inflammatory cells locally in the peritoneal cavity without inducing a systemic effect, as no changes in the number of inflammatory cells could be detected in blood or in the bone marrow. Collectively, these data suggest that this peptide could be a promising tool in the treatment of inflammatory diseases, such as inflammatory skin diseases, as it could reduce the number of inflammatory cells locally without suppressing the ability to combat infections.

3.
J Antibiot (Tokyo) ; 76(10): 603-612, 2023 10.
Article in English | MEDLINE | ID: mdl-37337088

ABSTRACT

Currently, antibiotic-resistant bacteria represent a serious threat to public health worldwide. Biofilm formation potentiates both virulence and antibiotic resistance of bacteria. Therefore, the discovery of new antibacterial and antibiofilm compounds is an issue of paramount importance to combat and prevent hard-to-treat bacterial infections. Zeolitic-imidazolate-frameworks (ZIFs) are metallo-organic compounds known to have various interesting chemical and biological applications, including antibacterial properties. In this study, we synthesized ZIF-67 nanoparticles, formed by imidazolate anions and cobalt cations, and found that they inhibit the growth of Acinetobacter baumannii, Pseudomonas aeruginosa, and Staphylococcus aureus. Sub-inhibitory concentrations of ZIF-67 were also able to significantly reduce the biomass of pre-established biofilms of these pathogenic bacteria. On the other hand, the ZIF-67 nanoparticles had null or low cytotoxicity in mammalian cells at those concentrations showing antibacterial or antibiofilm activities. Thus, our results reveal the potential of ZIF-67 nanoparticles to be used against pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents , Staphylococcus aureus , Animals , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacteria , Biofilms , Mammals
5.
J Cell Physiol ; 236(9): 6643-6656, 2021 09.
Article in English | MEDLINE | ID: mdl-33533043

ABSTRACT

Aberrant expression of CD43 in malignant tumors of nonhematopoietic origin such as those from lung, cervix, colon, and breast has been shown to correlate with poor prognosis, providing tumor cells with enhanced motility, anchorage-independent growth, and in vivo tumor size, while protecting the cells of NK lysis and apoptosis. To further characterize the role of CD43 in cell transformation, we tested whether interfering its expression modified the capacity of the A549 non-small cell lung cancer cells to secrete molecules contributing to malignancy. The proteomic analysis of the secretome of serum-starved A549 cells revealed that cells expressing normal levels of CD43 released significantly high levels of molecules involved in extracellular matrix organization, angiogenesis, platelet degranulation, collagen degradation, and inflammation, as compared to CD43 RNAi cells. This data reveals a novel and unexpected role for CD43 in lung cancer development, mainly in remodeling the tumor microenvironment.


Subject(s)
Extracellular Matrix/metabolism , Leukosialin/metabolism , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , A549 Cells , Gene Silencing , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Tumor Microenvironment
6.
PLoS One ; 13(10): e0205727, 2018.
Article in English | MEDLINE | ID: mdl-30325956

ABSTRACT

The occurrence of nosocomial infections has been on the rise for the past twenty years. Notably, infections caused by the Gram-positive bacteria Staphylococcus aureus represent a major clinical problem, as an increase in antibiotic multi-resistant strains has accompanied this rise. There is thus a crucial need to find and characterize new antibiotics against Gram-positive bacteria, and against antibiotic-resistant strains in general. We identified a new dermaseptin, DMS-DA6, produced by the skin of the Mexican frog Pachymedusa dacnicolor, with specific antibacterial activity against Gram-positive bacteria. This peptide is particularly effective against two multiple drug-resistant strains Enterococcus faecium BM4147 and Staphylococcus aureus DAR5829, and has no hemolytic activity. DMS-DA6 is naturally produced with the C-terminal carboxyl group in either the free or amide forms. By using Gram-positive model membranes and different experimental approaches, we showed that both forms of the peptide adopt an α-helical fold and have the same ability to insert into, and to disorganize a membrane composed of anionic lipids. However, the bactericidal capacity of DMS-DA6-NH2 was consistently more potent than that of DMS-DA6-OH. Remarkably, rather than resulting from the interaction with the negatively charged lipids of the membrane, or from a more stable conformation towards proteolysis, the increased capacity to permeabilize the membrane of Gram-positive bacteria of the carboxyamidated form of DMS-DA6 was found to result from its enhanced ability to interact with peptidoglycan.


Subject(s)
Amphibian Proteins/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Anura/metabolism , Enterococcus faecium/drug effects , Membranes/drug effects , Peptidoglycan/pharmacology , Skin/chemistry , Staphylococcus aureus/drug effects , A549 Cells/drug effects , Amphibian Proteins/genetics , Amphibian Proteins/isolation & purification , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/isolation & purification , Circular Dichroism , Drug Synergism , Humans , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests
7.
Nat Commun ; 8(1): 428, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874660

ABSTRACT

Preterm birth places infants in an adverse environment that leads to abnormal brain development and cerebral injury through a poorly understood mechanism known to involve neuroinflammation. In this study, we integrate human and mouse molecular and neuroimaging data to investigate the role of microglia in preterm white matter damage. Using a mouse model where encephalopathy of prematurity is induced by systemic interleukin-1ß administration, we undertake gene network analysis of the microglial transcriptomic response to injury, extend this by analysis of protein-protein interactions, transcription factors and human brain gene expression, and translate findings to living infants using imaging genomics. We show that DLG4 (PSD95) protein is synthesised by microglia in immature mouse and human, developmentally regulated, and modulated by inflammation; DLG4 is a hub protein in the microglial inflammatory response; and genetic variation in DLG4 is associated with structural differences in the preterm infant brain. DLG4 is thus apparently involved in brain development and impacts inter-individual susceptibility to injury after preterm birth.Inflammation mediated by microglia plays a key role in brain injury associated with preterm birth, but little is known about the microglial response in preterm infants. Here, the authors integrate molecular and imaging data from animal models and preterm infants, and find that microglial expression of DLG4 plays a role.


Subject(s)
Disks Large Homolog 4 Protein/metabolism , Genomics , Infant, Premature/growth & development , Infant, Premature/metabolism , Microglia/metabolism , White Matter/growth & development , White Matter/metabolism , Animals , Brain/growth & development , Brain/metabolism , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Humans , Infant, Newborn , Inflammation/pathology , Interleukin-1beta/pharmacology , Magnetic Resonance Imaging , Mice , Microglia/drug effects , Neuropsychiatry , Protein Interaction Maps/genetics , Quantitative Trait Loci/genetics , STAT3 Transcription Factor/metabolism , Transcriptome/genetics
8.
FASEB J ; 30(6): 2370-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26979087

ABSTRACT

CC chemokine receptor type 2 (CCR2) is a key molecule in inflammatory diseases and is an obvious drug target for the treatment of inflammation. A number of nonpeptidic, competitive CCR2 antagonists have been developed, but none has yet been approved for clinical use. Our aim was to identify a short peptide that showed allosteric antagonism against human and mouse CCR2. On the basis of sequence analysis and 3-dimensional modeling, we identified an original 7-d-amino acid peptidic CCR2 inhibitor that we have called extracellular loop 1 inverso (ECL1i), d(LGTFLKC). In vitro, ECL1i selectively and potently inhibits CC chemokine ligand type 2 (CCL2)-triggered chemotaxis (IC50, 2 µM) but no other conventional CCL2-associated events. We used the classic competitive CCR2 antagonist, BMS22 {2-[(isopropylaminocarbonyl)amino]-N-[2-[[cis-2-[[4-(methylthio)benzoyl]amino]cyclohexyl]amino]-2-oxoethyl]-5-(trifluoromethyl)benzamide}, as positive control and inhibited CCL2-dependent chemotaxis with an IC50 of 18 nM. As negative control, we used a peptide with the same composition as ECL1i, but in a different sequence, d(FKLTLCG). In vivo, ECL1i (4 mg/kg) interfered with CCR2-positive cell recruitment and attenuated disease progression in experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. This study establishes ECL1i as the first allosteric inhibitor of CCR2 with functional selectivity. ECL1i is a promising new agent in therapeutic development, and it may, by its selective effect, increase our understanding of CCR2 signaling pathways and functions.-Auvynet, C., Baudesson de Chanville, C., Hermand, P., Dorgham, K., Piesse, C., Pouchy, C., Carlier, L., Poupel, L., Barthélémy, S., Felouzis, V., Lacombe, C., Sagan, S., Salomon, B., Deterre, P., Sennlaub, F., Combadière, C. ECL1i, d(LGTFLKC), a novel, small peptide that specifically inhibits CCL2-dependent migration.


Subject(s)
Cell Movement/physiology , Chemokine CCL2/metabolism , Oligopeptides/pharmacology , Receptors, CCR2/metabolism , Animals , CHO Cells , Chemokine CCL2/genetics , Cricetulus , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Gene Expression Regulation/physiology , Humans , Male , Mice , Mice, Knockout , Models, Molecular , Oligopeptides/chemistry , Protein Binding , Protein Conformation , Receptors, CCR2/genetics
9.
J Med Chem ; 58(3): 1089-99, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25587631

ABSTRACT

Recruitment of leukocytes is essential to fight infections or to heal injuries; however, excessive and/or prolonged responses favor the development of major inflammatory pathologies, such as cardiovascular or neurodegenerative diseases. Thus, it is of great interest to seek novel compounds that can regulate leukocyte recruitment depending on the degree of inflammation. We have isolated and characterized, by different chromatographic techniques, mass spectrometry, and Edman sequencing, a new hexapeptide (SSLSKL) from the Mexican frog Pachymedusa dacnicolor, which we named pachymodulin. In vitro, pachymodulin promotes the migration of leukocytes through the binding and activation of the human and mouse N-formyl peptide receptor 2 (huFPR2). In vivo, it exhibits opposite biological activities: under homeostatic conditions, pachymodulin induces the recruitment of leukocytes, whereas under inflammatory conditions, it inhibits this process. Therefore, pachymodulin represents an interesting template in the quest to design new immunomodulatory drugs in the therapy of immune-related diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunologic Factors/pharmacology , Oligopeptides/pharmacology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Skin/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Anura , Cell Movement/drug effects , Dose-Response Relationship, Drug , Humans , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Inflammation/drug therapy , Leukocytes/drug effects , Ligands , Mice , Molecular Structure , Oligopeptides/chemistry , Oligopeptides/isolation & purification , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Structure-Activity Relationship
10.
Brain Behav Immun ; 45: 198-210, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25449579

ABSTRACT

Neuropathic pain resulting from peripheral nerve injury involves many persistent neuroinflammatory processes including inflammatory chemokines that control leukocyte trafficking and activate resident cells. Several studies have shown that CCL2 chemokine, a potent attractant of monocytes, and its cognate receptor, CCR2, play a critical role in regulating nociceptive processes during neuropathic pain. However, the role of CCL2 in peripheral leukocyte infiltration-associated neuropathic pain remains poorly understood. In particular, the contribution of individual CCL2-expressing cell populations (i.e. stromal and leukocytes) to immune cell recruitment into the injured nerve has not been established. Here, in preclinical model of peripheral neuropathic pain (i.e. chronic constriction injury of the sciatic nerve), we have demonstrated that, CCL2 content was increased specifically in nerve fibers. This upregulation of CCL2 correlated with local monocyte/macrophage infiltration and pain processing. Furthermore, sciatic intraneural microinjection of CCL2 in naïve animals triggered long-lasting pain behavior associated with local monocyte/macrophage recruitment. Using a specific CCR2 antagonist and mice with a CCL2 genetic deletion, we have also established that the CCL2/CCR2 axis drives monocyte/macrophage infiltration and pain hypersensitivity in the CCI model. Finally, specific deletion of CCL2 in stromal or immune cells respectively using irradiated bone marrow-chimeric CCI mice demonstrated that stromal cell-derived CCL2 (in contrast to CCL2 immune cell-derived) tightly controls monocyte/macrophage recruitment into the lesion and plays a major role in the development of neuropathic pain. These findings demonstrate that in chronic pain states, CCL2 expressed by sciatic nerve cells predominantly drove local neuro-immune interactions and pain-related behavior through CCR2 signaling.


Subject(s)
Chemokine CCL2/immunology , Macrophages/immunology , Monocytes/immunology , Neuralgia/immunology , Peripheral Nerve Injuries/immunology , Sciatic Nerve/injuries , Animals , Bone Marrow Transplantation , Constriction, Pathologic , Hyperalgesia/genetics , Hyperalgesia/immunology , Mice , Myeloid Cells/immunology , Rats , Sciatic Nerve/immunology , Up-Regulation
11.
EMBO Mol Med ; 5(11): 1775-93, 2013 11.
Article in English | MEDLINE | ID: mdl-24142887

ABSTRACT

Atrophic age-related macular degeneration (AMD) is associated with the subretinal accumulation of mononuclear phagocytes (MPs). Their role in promoting or inhibiting retinal degeneration is unknown. We here show that atrophic AMD is associated with increased intraocular CCL2 levels and subretinal CCR2(+) inflammatory monocyte infiltration in patients. Using age- and light-induced subretinal inflammation and photoreceptor degeneration in Cx3cr1 knockout mice, we show that subretinal Cx3cr1 deficient MPs overexpress CCL2 and that both the genetic deletion of CCL2 or CCR2 and the pharmacological inhibition of CCR2 prevent inflammatory monocyte recruitment, MP accumulation and photoreceptor degeneration in vivo. Our study shows that contrary to CCR2 and CCL2, CX3CR1 is constitutively expressed in the retina where it represses the expression of CCL2 and the recruitment of neurotoxic inflammatory CCR2(+) monocytes. CCL2/CCR2 inhibition might represent a powerful tool for controlling inflammation and neurodegeneration in AMD.


Subject(s)
Macular Degeneration/immunology , Monocytes/immunology , Photoreceptor Cells, Vertebrate/immunology , Receptors, CCR2/immunology , Receptors, Chemokine/deficiency , Animals , CX3C Chemokine Receptor 1 , Chemokine CCL2/immunology , Female , Humans , Inflammation/genetics , Inflammation/immunology , Macular Degeneration/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Receptors, CCR2/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology
12.
Arterioscler Thromb Vasc Biol ; 33(10): 2297-305, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23887641

ABSTRACT

OBJECTIVE: Alterations of the chemokine receptor CX3CR1 gene were associated with a reduced risk of myocardial infarction in human and limited atherosclerosis in mice. In this study, we addressed whether CX3CR1 antagonists are potential therapeutic tools to limit acute and chronic inflammatory processes in atherosclerosis. APPROACH AND RESULTS: Treatment with F1, an amino terminus-modified CX3CR1 ligand endowed with CX3CR1 antagonist activity, reduced the extent of atherosclerotic lesions in both Apoe(-/-) and Ldlr(-/-) proatherogenic mouse models. Macrophage accumulation in the aortic sinus was reduced in F1-treated Apoe(-/-) mice but the macrophage density of the lesions was similar in F1-treated and control mice. Both in vitro and in vivo F1 treatment reduced CX3CR1-dependent inflammatory monocyte adhesion, potentially limiting their recruitment. In addition, F1-treated Apoe(-/-) mice displayed reduced numbers of blood inflammatory monocytes, whereas resident monocyte numbers remained unchanged. Both in vitro and in vivo F1 treatment reduced CX3CR1-dependent inflammatory monocyte survival. Finally, F1 treatment of Apoe(-/-) mice with advanced atherosclerosis led to smaller lesions than untreated mice but without reverting to the initial phenotype. CONCLUSIONS: The CX3CR1 antagonist F1 is a potent inhibitor of the progression of atherosclerotic lesions by means of its selective impact on inflammatory monocyte functions. Controlling monocyte trafficking and survival may be an alternative or complementary therapy to lipid-lowering drugs classically used in the treatment of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Chemokine CX3CL1/pharmacology , Hypolipidemic Agents/pharmacology , Peptides/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , CX3C Chemokine Receptor 1 , Cells, Cultured , Disease Models, Animal , Humans , Ligands , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Receptors, Chemokine/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors
13.
Neoplasia ; 15(6): 641-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23730212

ABSTRACT

Expression of the CC chemokine receptor 1 (CCR1) by tumor cells has been associated with protumoral activity; however, its role in nontumoral cells during tumor development remains elusive. Here, we investigated the role of CCR1 deletion on stromal and hematopoietic cells in a liver metastasis tumor model. Metastasis development was strongly impaired in CCR1-deficient mice compared to control mice and was associated with reduced liver monocyte infiltration. To decipher the role of myeloid cells, sublethally irradiated mice were reconstituted with CCR1-deficient bone marrow (BM) and showed better survival rates than the control reconstituted mice. These results point toward the involvement of CCR1 myeloid cell infiltration in the promotion of tumor burden. In addition, survival rates were extended in CCR1-deficient mice receiving either control or CCR1-deficient BM, indicating that host CCR1 expression on nonhematopoietic cells also supports tumor growth. Finally, we found defective tumor-induced neoangiogenesis (in vitro and in vivo) in CCR1-deficient mice. Overall, our results indicate that CCR1 expression by both hematopoietic and nonhematopoietic cells favors tumor aggressiveness. We propose CCR1 as a potential therapeutical target for liver metastasis therapy.


Subject(s)
Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver/blood supply , Myeloid Cells/pathology , Receptors, CCR1/metabolism , Animals , Bone Marrow Transplantation , Female , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/therapy , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/pathology , Neovascularization, Pathologic , Receptors, CCR1/genetics , Survival Rate
14.
Glycobiology ; 23(1): 32-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22942212

ABSTRACT

An important step of innate immune response is the recruitment of polymorphonuclear leukocytes (PMN) to injured tissues through chemotactic molecules. Galectins, a family of endogenous lectins, participate in numerous functions such as lymphoid cell migration, homing, cell-cell and cell-matrix interactions. Particularly, galectin-3 (Gal-3) and -9 have been implicated in the modulation of acute and chronic inflammation by inducing the directional migration of monocytes/macrophages and eosinophils, whereas Gal-1 is considered to function as an anti-inflammatory molecule, capable of inhibiting the influx of PMN to the site of injury. In this study, we assessed the effect of Gal-1 on neutrophil recruitment, in the absence of additional inflammatory insults. Contrasting with its capacity to inhibit cell trafficking and modulate the release of mediators described in models of acute inflammation and autoimmunity, we evidenced that Gal-1 has the capacity to induce neutrophil migration both in vitro and in vivo. This effect is not mediated through a G-protein-coupled receptor but potentially through the sialoglycoprotein CD43, via carbohydrate binding and through the p38 mitogen-activated protein kinase pathway. These results suggest a novel biological function for CD43 on neutrophils and highlight that depending on the environment, Gal-1 can act either as chemoattractant or, as a molecule that negatively regulates migration under acute inflammatory conditions, underscoring the potential of Gal-1 as a target for innovative drug development.


Subject(s)
Chemotaxis, Leukocyte , Galectin 1/metabolism , Neutrophils/physiology , Galectin 1/pharmacology , Humans , Immunity, Innate , In Vitro Techniques , Leukosialin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Arterioscler Thromb Vasc Biol ; 32(9): 2206-13, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22796582

ABSTRACT

OBJECTIVE: Leukocyte infiltration in ischemic areas is a hallmark of myocardial infarction, and overwhelming infiltration of innate immune cells has been shown to promote adverse remodeling and cardiac rupture. Recruitment of inflammatory cells in the ischemic heart depends highly on the family of CC-chemokines and their receptors. Here, we hypothesized that the chemokine decoy receptor D6, which specifically binds and scavenges inflammatory CC-chemokines, might limit inflammation and adverse cardiac remodeling after infarction. METHODS AND RESULTS: D6 was expressed in human and murine infarcted myocardium. In a murine model of myocardial infarction, D6 deficiency led to increased chemokine (C-C motif) ligand 2 and chemokine (C-C motif) ligand 3 levels in the ischemic heart. D6-deficient (D6(-/-)) infarcts displayed increased infiltration of pathogenic neutrophils and Ly6Chi monocytes, associated with strong matrix metalloproteinase-9 and matrix metalloproteinase-2 activities in the ischemic heart. D6(-/-) mice were cardiac rupture prone after myocardial infarction, and functional analysis revealed that D6(-/-) hearts had features of adverse remodeling with left ventricle dilation and reduced ejection fraction. Bone marrow chimera experiments showed that leukocyte-borne D6 had no role in this setting, and that leukocyte-specific chemokine (C-C motif) receptor 2 deficiency rescued the adverse phenotype observed in D6(-/-) mice. CONCLUSIONS: We show for the first time that the chemokine decoy receptor D6 limits CC-chemokine-dependent pathogenic inflammation and is required for adequate cardiac remodeling after myocardial infarction.


Subject(s)
Inflammation/prevention & control , Myocardial Infarction/immunology , Myocardium/immunology , Receptors, CCR10/metabolism , Receptors, Chemokine/metabolism , Ventricular Remodeling , Animals , Antigens, Ly/metabolism , Bone Marrow Transplantation , Chemokine CCL2/metabolism , Chemokine CCL3/metabolism , Chemotaxis , Disease Models, Animal , Genotype , Heart Rupture, Post-Infarction/immunology , Heart Rupture, Post-Infarction/pathology , Humans , Hypertrophy, Left Ventricular/immunology , Hypertrophy, Left Ventricular/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Myocardial Infarction/complications , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Neutrophil Infiltration , Neutrophils/immunology , Phenotype , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Signal Transduction , Stroke Volume , Ultrasonography , Ventricular Function, Left , Chemokine Receptor D6
16.
Am J Pathol ; 180(3): 1040-1048, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22203055

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli is the main etiological agent that causes hemolytic uremic syndrome (HUS), a microangiopathic disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure. Although direct cytotoxic effects on endothelial cells by Stx are the primary pathogenic event, there is evidence that indicates the inflammatory response mediated by polymorphonuclear neutrophils and monocytes as the key event during HUS development. Because the chemokine receptor CCR1 participates in the pathogenesis of several renal diseases by orchestrating myeloid cell kidney infiltration, we specifically addressed the contribution of CCR1 in a murine model of HUS. We showed that Stx type 2-treated CCR1(-/-) mice have an increased survival rate associated with less functional and histological renal damage compared with control mice. Stx type 2-triggered neutrophilia and monocytosis and polymorphonuclear neutrophil and monocyte renal infiltration were significantly reduced and delayed in CCR1(-/-) mice compared with control mice. In addition, the increase of the inflammatory cytokines (tumor necrosis factor-α and IL-6) in plasma was delayed in CCR1(-/-) mice compared with control mice. These data demonstrate that CCR1 participates in cell recruitment to the kidney and amplification of the inflammatory response that contributes to HUS development. Blockade of CCR1 could be important to the design of future therapies to restrain the inflammatory response involved in the development of HUS.


Subject(s)
Hemolytic-Uremic Syndrome/chemically induced , Receptors, CCR1/physiology , Shiga Toxin 2/toxicity , Animals , Bone Marrow/pathology , Creatine/metabolism , Hemolytic-Uremic Syndrome/pathology , Interleukin-6/metabolism , Kidney Tubules/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Myeloid Cells/physiology , Receptors, CCR1/deficiency , Survival Rate , Tumor Necrosis Factor-alpha/metabolism , Urea/metabolism
17.
J Neuroinflammation ; 7: 87, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-21126357

ABSTRACT

The causes of age-related macular degeneration (AMD) are not well understood. Due to demographic shifts in the industrialized world a growing number of people will develop AMD in the coming decades. To develop treatments it is essential to characterize the disease's pathogenic process. Over the past few years, numerous studies have focused on the role of chemotactic cytokines, also known as chemokines. Certain chemokines, such as CCL2 and CX3CL1, appear to be crucial in subretinal microglia and macrophage accumulation observed in AMD, and participate in the development of retinal degeneration as well as in choroidal neovascularization. This paper reviews the possible implications of CCL2 and CX3CL1 signaling in AMD. Expression patterns, single nucleotide polymorphisms (SNPs) association studies, chemokine and chemokine receptor knockout models are discussed. Future AMD treatments could target chemokines and/or their receptors.


Subject(s)
Chemokine CCL2/immunology , Chemokine CX3CL1/immunology , Macular Degeneration/immunology , Receptors, CCR2/immunology , Animals , CX3C Chemokine Receptor 1 , Chemokine CCL2/genetics , Chemokine CX3CL1/genetics , Disease Models, Animal , Humans , Macular Degeneration/pathology , Macular Degeneration/physiopathology , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Receptors, CCR2/genetics , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Retina/cytology , Retina/immunology , Signal Transduction/physiology
18.
Biol Aujourdhui ; 204(4): 311-9, 2010.
Article in French | MEDLINE | ID: mdl-21215248

ABSTRACT

Age-related macular degeneration (AMD) is the main cause of irreversible blindness in industrialized nations. Recent research has emphasized the importance of inflammatory processes in pathogenesis of this disease. Chemotactic cytokines also named chemokines are important mediators of inflammation and might have a role in development of this disease. They appear to be crucial in the subretinal microglia / macrophage accumulation observed in AMD and may participate in the development of retinal degeneration and in choroidal neovascularization. This paper reviews the possible implication of chemokines in the development of AMD.


Subject(s)
Chemokines/physiology , Macular Degeneration/immunology , Animals , CX3C Chemokine Receptor 1 , Chemokine CCL2/physiology , Humans , Models, Immunological , Receptors, Chemokine/physiology
19.
FEBS J ; 276(22): 6773-86, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19843179

ABSTRACT

Antimicrobial peptides participate in innate host defense by directly eliminating pathogens as a result of their ability to damage the microbial membrane and by providing danger signals that will recruit innate immune cells to the site of infection. Dermaseptin DA4 (DRS-DA4), a new antimicrobial peptide of the dermaseptin superfamily, was identified based on its chemotactic properties, contrasting with the currently used microbicidal properties assessment. The peptide was isolated and purified by size exclusion HPLC and RP-HPLC from the skin of the Mexican frog, Pachymedusa dacnicolor. MS and amino acid sequence analyses were consistent with the structure GMWSKIKNAGKAAKAAAKAAGKAALGAVSEAM. CD experiments showed that, unlike most antimicrobial peptides of the dermaseptin superfamily, DRS-DA4 is not structured in the presence of zwitterionic lipids. DRS-DA4 is a potent chemoattractant for human leukocytes and is devoid of hemolytic activity; in addition, bactericidal tests and membrane perturbation assays on model membranes and on Escherichia coli and Staphylococcus aureus strains have shown that the antibacterial effects of DRS-DA4 and permeabilization of the inner membrane are exclusively selective for Gram-negative bacteria. Interestingly, despite high sequence homology with dermaseptin S4, dermaseptin B2 was not able to induce directional migration of leukocytes, and displayed a broader bactericidal spectrum. A detailed structure-function analysis of closely related peptides with different capabilities, such as DRS-DA4 and dermaseptin B2, is critical for the design of new molecules with specific attributes to modulate immunity and/or act as microbicidal agents.


Subject(s)
Amphibian Proteins/pharmacology , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Chemotactic Factors/pharmacology , Gram-Negative Bacteria/drug effects , Amino Acid Sequence , Amphibian Proteins/chemistry , Animals , Antimicrobial Cationic Peptides/chemistry , Anura/metabolism , Circular Dichroism , Humans , Leukocytes/drug effects , Microbial Sensitivity Tests , Molecular Sequence Data , Monocytes/drug effects , Neutrophils/drug effects , Receptors, G-Protein-Coupled/metabolism , Sequence Homology, Amino Acid , Skin/metabolism
20.
FEBS J ; 276(22): 6497-508, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19817855

ABSTRACT

The term 'antimicrobial peptides' refers to a large number of peptides first characterized on the basis of their antibiotic and antifungal activities. In addition to their role as endogenous antibiotics, antimicrobial peptides, also called host defense peptides, participate in multiple aspects of immunity (inflammation, wound repair, and regulation of the adaptive immune system) as well as in maintaining homeostasis. The possibility of utilizing these multifunctional molecules to effectively combat the ever-growing group of antibiotic-resistant pathogens has intensified research aimed at improving their antibiotic activity and therapeutic potential, without the burden of an exacerbated inflammatory response, but conserving their immunomodulatory potential. In this minireview, we focus on the contribution of small cationic antimicrobial peptides - particularly human cathelicidins and defensins - to the immune response and disease, highlighting recent advances in our understanding of the roles of these multifunctional molecules.


Subject(s)
Adaptive Immunity/immunology , Antimicrobial Cationic Peptides/immunology , Immunity, Innate/immunology , Animals , Antimicrobial Cationic Peptides/metabolism , Cathelicidins/immunology , Cathelicidins/metabolism , Defensins/immunology , Defensins/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...